Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
1.
Oncologist ; 25(9): 810-817, 2020 09.
Article in English | MEDLINE | ID: mdl-32372416

ABSTRACT

PURPOSE: Delta-like protein 3 (DLL3) is being developed as a predictive biomarker for DLL3-targeting antibody-drug conjugate and other therapies. Given the neuroendocrine features of Merkel cell carcinoma (MCC), we sought to evaluate DLL3 expression and its role in MCC. EXPERIMENTAL DESIGN: Formalin-fixed and paraffin-embedded MCC cases were consecutively selected. Immunohistochemistry was performed for DLL3 (SC16.65 antibody) and polyomavirus large T-antigen (sc-136172 antibody). Slides were read out for percentage of positive tumor cells. Cox proportional hazards model was applied to assess the association between DLL3 expression and overall survival (OS). A patient with a DLL3-expressing MCC was treated with rovalpituzumab tesirine (Rova-T) in the "other tumor" cohort of NCT02709889 and assessed for response. RESULTS: The median H-score of DLL3 expression of 65 patients included was 60 (interquartile range, 30-100). Fifty-eight cases (89%) had ≥1% tumor cells positive for DLL3 expression with any intensity, of which the median DLL3 expression was 50% (interquartile range, 25%-70%). Thirty-four cases (52%) had ≥50% tumor cells positive for DLL3 expression with any intensity. Higher H-score of DLL3 expression was associated with higher polyomavirus nuclear expression (p = .003) when it was dichotomized to negative versus positive. H-score of DLL3 expression did not predict OS of patients with MCC (p = .4) after being adjusted for common clinicopathological factors. A patient treated with Rova-T for refractory metastatic MCC achieved partial response. CONCLUSIONS: DLL3 overexpression is very common in MCC by immunohistochemistry. The response to treatment suggests that DLL3 expression may have predictive relevance for DLL3-targeting therapies in MCC. IMPLICATIONS FOR PRACTICE: Delta-like protein 3 (DLL3) is being developed as a predictive biomarker to identify patients for treatment with DLL3-targeting agents. Merkel cell carcinoma (MCC) is an aggressive neuroendocrine carcinoma of the skin. It was found that DLL3 overexpression is very common in MCC by immunohistochemistry and significantly associated with Merkel cell polyomavirus expression. Despite the lack of prognostic significance in this cohort, DLL3 expression may have predictive relevance for DLL3-targeting therapies in MCC. The high levels of DLL3 expression in a subset of MCC may potentially be used to select patients to receive DLL3-targeting therapies.


Subject(s)
Carcinoma, Merkel Cell , Skin Neoplasms , Carcinoma, Merkel Cell/drug therapy , Carcinoma, Merkel Cell/genetics , Humans , Intracellular Signaling Peptides and Proteins/genetics , Membrane Proteins/genetics , Skin Neoplasms/drug therapy , Skin Neoplasms/genetics
2.
Proc Natl Acad Sci U S A ; 115(44): 11244-11249, 2018 10 30.
Article in English | MEDLINE | ID: mdl-30322923

ABSTRACT

The E3 ubiquitin ligase CRL4COP1/DET1 is active in the absence of ERK signaling, modifying the transcription factors ETV1, ETV4, ETV5, and c-JUN with polyubiquitin that targets them for proteasomal degradation. Here we show that this posttranslational regulatory mechanism is active in neurons, with ETV5 and c-JUN accumulating within minutes of ERK activation. Mice with constitutive photomorphogenesis 1 (Cop1) deleted in neural stem cells showed abnormally elevated expression of ETV1, ETV4, ETV5, and c-JUN in the developing brain and spinal cord. Expression of c-JUN target genes Vimentin and Gfap was increased, whereas ETV5 and c-JUN both contributed to an expanded number of cells expressing genes associated with gliogenesis, including Olig1, Olig2, and Sox10. The mice had subtle morphological abnormalities in the cerebral cortex, hippocampus, and cerebellum by embryonic day 18 and died soon after birth. Elevated c-JUN, ETV5, and ETV1 contributed to the perinatal lethality, as several Cop1-deficient mice also lacking c-Jun and Etv5, or lacking Etv5 and heterozygous for Etv1, were viable.


Subject(s)
Brain/metabolism , Nuclear Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , Ubiquitin/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , DNA-Binding Proteins/metabolism , Male , Mice , Mice, Inbred C57BL , Proto-Oncogene Proteins c-ets/metabolism , Proto-Oncogene Proteins c-jun/metabolism , Transcription Factors/metabolism
3.
J Clin Invest ; 128(10): 4485-4500, 2018 10 01.
Article in English | MEDLINE | ID: mdl-30024858

ABSTRACT

Oxidative stress is an underlying component of acute and chronic kidney disease. Apoptosis signal-regulating kinase 1 (ASK1) is a widely expressed redox-sensitive serine threonine kinase that activates p38 and c-Jun N-terminal kinase (JNK) mitogen-activated protein kinase kinases, and induces apoptotic, inflammatory, and fibrotic signaling in settings of oxidative stress. We describe the discovery and characterization of a potent and selective small-molecule inhibitor of ASK1, GS-444217, and demonstrate the therapeutic potential of ASK1 inhibition to reduce kidney injury and fibrosis. Activation of the ASK1 pathway in glomerular and tubular compartments was confirmed in renal biopsies from patients with diabetic kidney disease (DKD) and was decreased by GS-444217 in several rodent models of kidney injury and fibrosis that collectively represented the hallmarks of DKD pathology. Treatment with GS-444217 reduced progressive inflammation and fibrosis in the kidney and halted glomerular filtration rate decline. Combination of GS-444217 with enalapril, an angiotensin-converting enzyme inhibitor, led to a greater reduction in proteinuria and regression of glomerulosclerosis. These results identify ASK1 as an important target for renal disease and support the clinical development of an ASK1 inhibitor for the treatment of DKD.


Subject(s)
Diabetic Nephropathies/enzymology , Fibroblasts/enzymology , Kidney Glomerulus/enzymology , MAP Kinase Kinase Kinase 5/metabolism , MAP Kinase Signaling System , Animals , Diabetic Nephropathies/drug therapy , Diabetic Nephropathies/genetics , Diabetic Nephropathies/pathology , Disease Models, Animal , Female , Fibroblasts/pathology , Fibrosis , Humans , Kidney Glomerulus/pathology , MAP Kinase Kinase Kinase 5/antagonists & inhibitors , MAP Kinase Kinase Kinase 5/genetics , Male , Mice , Mice, Knockout , Protein Kinase Inhibitors/pharmacology , Random Allocation , Rats, Sprague-Dawley
4.
J Hepatol ; 68(5): 912-921, 2018 05.
Article in English | MEDLINE | ID: mdl-29247724

ABSTRACT

BACKGROUND & AIMS: GS-9620, an oral agonist of toll-like receptor 7, is in clinical development for the treatment of chronic hepatitis B (CHB). GS-9620 was previously shown to induce prolonged suppression of serum viral DNA and antigens in the chimpanzee and woodchuck models of CHB. Herein, we investigated the immunomodulatory mechanisms underlying these antiviral effects. METHODS: Archived liver biopsies and paired peripheral blood mononuclear cell samples from a previous chimpanzee study were analyzed by RNA sequencing, quantitative reverse transcription PCR, immunohistochemistry (IHC) and in situ hybridization (ISH). RESULTS: GS-9620 treatment of CHB chimpanzees induced an intrahepatic transcriptional profile significantly enriched with genes associated with hepatitis B virus (HBV) clearance in acutely infected chimpanzees. Type I and II interferon, CD8+ T cell and B cell transcriptional signatures were associated with treatment response, together with evidence of hepatocyte death and liver regeneration. IHC and ISH confirmed an increase in intrahepatic CD8+ T cell and B cell numbers during treatment, and revealed that GS-9620 transiently induced aggregates predominantly comprised of CD8+ T cells and B cells in portal regions. There were no follicular dendritic cells or IgG-positive cells in these lymphoid aggregates and very few CD11b+ myeloid cells. There was no change in intrahepatic natural killer cell number during GS-9620 treatment. CONCLUSION: The antiviral response to GS-9620 treatment in CHB chimpanzees was associated with an intrahepatic interferon response and formation of lymphoid aggregates in the liver. Our data indicate these intrahepatic structures are not fully differentiated follicles containing germinal center reactions. However, the temporal correlation between development of these T and B cell aggregates and the antiviral response to treatment suggests they play a role in promoting an effective immune response against HBV. LAY SUMMARY: New therapies to treat chronic hepatitis B (CHB) are urgently needed. In this study we performed a retrospective analysis of liver and blood samples from a chimpanzee model of CHB to help understand how GS-9620, a drug in clinical trials, suppressed hepatitis B virus (HBV). We found that the antiviral response to GS-9620 was associated with accumulation of immune cells in the liver that can either kill cells infected with HBV or can produce antibodies that may prevent HBV from infecting new liver cells. These findings have important implications for how GS-9620 may be used in patients and may also help guide the development of new therapies to treat chronic HBV infection.


Subject(s)
Antiviral Agents/pharmacology , Hepatitis B, Chronic/drug therapy , Hepatitis B, Chronic/immunology , Pteridines/pharmacology , Toll-Like Receptor 7/agonists , Animals , B-Lymphocytes/drug effects , B-Lymphocytes/immunology , B-Lymphocytes/pathology , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/pathology , Cell Aggregation/drug effects , Cell Aggregation/immunology , Disease Models, Animal , Gene Expression Profiling , Hepatitis B, Chronic/virology , Humans , Liver/drug effects , Liver/immunology , Liver/pathology , Pan troglodytes
5.
J Histochem Cytochem ; 63(4): 244-55, 2015 04.
Article in English | MEDLINE | ID: mdl-25575566

ABSTRACT

With recent advances in immunohistochemical (IHC) techniques, immunohistochemistry now plays a more important role in research, especially in mouse models where characterization of cellular patterns of protein expression has become critical. Even with these recent advances, a paucity of IHC quality antibodies for some proteins still exists. To address this, we have developed a novel IHC assay that utilizes a commercially available goat anti-DDDDK peptide polyclonal antibody on paraffin-embedded tissues from knock-in mice expressing proteins of interest tagged with a 3 × FLAG epitope at physiologically relevant levels. Focusing on two 3 × FLAG-tagged proteins for which specific antibodies were available, USP48 and RIPK3, we were able to validate our anti-DDDDK assay by comparing the IHC directed against the actual proteins to the anti-DDDDK IHC assay, which recognizes the FLAG epitope. We were also able to detect a third 3 × FLAG-tagged protein, BAP1, for which quality reagents were not available. This universal IHC method will enable researchers to characterize the expression patterns of proteins of interest when specific antibodies are lacking.


Subject(s)
Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Tumor Suppressor Proteins/metabolism , Ubiquitin Thiolesterase/metabolism , Ubiquitin-Specific Proteases/metabolism , Animals , Antibodies , Epitopes , Gene Knock-In Techniques , Goats , Immunohistochemistry , Mice, Inbred C57BL , Mice, Mutant Strains , Oligopeptides/immunology , Oligopeptides/metabolism , Organ Specificity , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Tumor Suppressor Proteins/genetics , Ubiquitin Thiolesterase/genetics , Ubiquitin-Specific Proteases/genetics
6.
Hepatology ; 61(3): 942-52, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25311838

ABSTRACT

UNLABELLED: Primary liver cancer encompasses both hepatocellular carcinoma (HCC) and cholangiocarcinoma (CCA). The Notch signaling pathway, known to be important for the proper development of liver architecture, is also a potential driver of primary liver cancer. However, with four known Notch receptors and several Notch ligands, it is not clear which Notch pathway members play the predominant role in liver cancer. To address this question, we utilized antibodies to specifically target Notch1, Notch2, Notch3, or jagged1 (Jag1) in a mouse model of primary liver cancer driven by v-akt murine thymoma viral oncogene homolog and neuroblastoma RAS viral oncogene homolog (NRas). We show that inhibition of Notch2 reduces tumor burden by eliminating highly malignant HCC- and CCA-like tumors. Inhibition of the Notch ligand, Jag1, had a similar effect, consistent with Jag1 acting in cooperation with Notch2. This effect was specific to Notch2, because Notch3 inhibition did not decrease tumor burden. Unexpectedly, Notch1 inhibition altered the relative proportion of tumor types, reducing HCC-like tumors but dramatically increasing CC-like tumors. Finally, we show that Notch2 and Jag1 are expressed in, and Notch2 signaling is activated in, a subset of human HCC samples. CONCLUSIONS: These findings underscore the distinct roles of different Notch receptors in the liver and suggest that inhibition of Notch2 signaling represents a novel therapeutic option in the treatment of liver cancer.


Subject(s)
Liver Neoplasms/drug therapy , Receptors, Notch/antagonists & inhibitors , Animals , Calcium-Binding Proteins/analysis , Calcium-Binding Proteins/antagonists & inhibitors , Disease Models, Animal , Forkhead Box Protein M1 , Forkhead Transcription Factors/analysis , Humans , Intercellular Signaling Peptides and Proteins/analysis , Jagged-1 Protein , Membrane Proteins/analysis , Membrane Proteins/antagonists & inhibitors , Mice , Proto-Oncogene Proteins c-akt/physiology , Proto-Oncogene Proteins p21(ras)/physiology , Receptors, Notch/analysis , Receptors, Notch/physiology , Serrate-Jagged Proteins
7.
Mol Cancer Ther ; 13(11): 2630-40, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25249555

ABSTRACT

Mesothelin (MSLN) is an attractive target for antibody-drug conjugate therapy because it is highly expressed in various epithelial cancers, with normal expression limited to nondividing mesothelia. We generated novel antimesothelin antibodies and conjugated an internalizing one (7D9) to the microtubule-disrupting drugs monomethyl auristatin E (MMAE) and MMAF, finding the most effective to be MMAE with a lysosomal protease-cleavable valine-citrulline linker. The humanized (h7D9.v3) version, αMSLN-MMAE, specifically targeted mesothelin-expressing cells and inhibited their proliferation with an IC50 of 0.3 nmol/L. Because the antitumor activity of an antimesothelin immunotoxin (SS1P) in transfected mesothelin models did not translate to the clinic, we carefully selected in vivo efficacy models endogenously expressing clinically relevant levels of mesothelin, after scoring mesothelin levels in ovarian, pancreatic, and mesothelioma tumors by immunohistochemistry. We found that endogenous mesothelin in cancer cells is upregulated in vivo and identified two suitable xenograft models for each of these three indications. A single dose of αMSLN-MMAE profoundly inhibited or regressed tumor growth in a dose-dependent manner in all six models, including two patient-derived tumor xenografts. The robust and durable efficacy of αMSLN-MMAE in preclinical models of ovarian, mesothelioma, and pancreatic cancers justifies the ongoing phase I clinical trial.


Subject(s)
GPI-Linked Proteins/immunology , Immunotoxins/pharmacology , Mesothelioma/drug therapy , Oligopeptides/pharmacology , Ovarian Neoplasms/drug therapy , Pancreatic Neoplasms/drug therapy , Animals , Female , GPI-Linked Proteins/biosynthesis , Humans , Immunohistochemistry , Immunotoxins/chemistry , Immunotoxins/immunology , Mesothelin , Mice , Oligopeptides/chemistry , Random Allocation , Transfection , Xenograft Model Antitumor Assays
8.
Genome Biol ; 15(8): 436, 2014 Aug 26.
Article in English | MEDLINE | ID: mdl-25159915

ABSTRACT

BACKGROUND: Hepatocellular carcinoma (HCC) is a heterogeneous disease with high mortality rate. Recent genomic studies have identified TP53, AXIN1, and CTNNB1 as the most frequently mutated genes. Lower frequency mutations have been reported in ARID1A, ARID2 and JAK1. In addition, hepatitis B virus (HBV) integrations into the human genome have been associated with HCC. RESULTS: Here, we deep-sequence 42 HCC patients with a combination of whole genome, exome and transcriptome sequencing to identify the mutational landscape of HCC using a reasonably large discovery cohort. We find frequent mutations in TP53, CTNNB1 and AXIN1, and rare but likely functional mutations in BAP1 and IDH1. Besides frequent hepatitis B virus integrations at TERT, we identify translocations at the boundaries of TERT. A novel deletion is identified in CTNNB1 in a region that is heavily mutated in multiple cancers. We also find multiple high-allelic frequency mutations in the extracellular matrix protein LAMA2. Lower expression levels of LAMA2 correlate with a proliferative signature, and predict poor survival and higher chance of cancer recurrence in HCC patients, suggesting an important role of the extracellular matrix and cell adhesion in tumor progression of a subgroup of HCC patients. CONCLUSIONS: The heterogeneous disease of HCC features diverse modes of genomic alteration. In addition to common point mutations, structural variations and methylation changes, there are several virus-associated changes, including gene disruption or activation, formation of chimeric viral-human transcripts, and DNA copy number changes. Such a multitude of genomic events likely contributes to the heterogeneous nature of HCC.


Subject(s)
Carcinoma, Hepatocellular/genetics , DNA Mutational Analysis/methods , Genetic Variation , Laminin/genetics , Liver Neoplasms/genetics , Carcinoma, Hepatocellular/virology , Genetic Heterogeneity , Hepatitis B/genetics , Hepatitis B virus/physiology , High-Throughput Nucleotide Sequencing , Humans , Liver Neoplasms/virology , Mutation Rate , Survival Analysis
9.
Science ; 343(6177): 1357-60, 2014 Mar 21.
Article in English | MEDLINE | ID: mdl-24557836

ABSTRACT

Receptor-interacting protein kinase 1 (RIPK1) and RIPK3 trigger pro-inflammatory cell death termed "necroptosis." Studies with RIPK3-deficient mice or the RIPK1 inhibitor necrostatin-1 suggest that necroptosis exacerbates pathology in many disease models. We engineered mice expressing catalytically inactive RIPK3 D161N or RIPK1 D138N to determine the need for the active kinase in the whole animal. Unexpectedly, RIPK3 D161N promoted lethal RIPK1- and caspase-8-dependent apoptosis. In contrast, mice expressing RIPK1 D138N were viable and, like RIPK3-deficient mice, resistant to tumor necrosis factor (TNF)-induced hypothermia. Cells expressing RIPK1 D138N were resistant to TNF-induced necroptosis, whereas TNF-induced signaling pathways promoting gene transcription were unperturbed. Our data indicate that the kinase activity of RIPK3 is essential for necroptosis but also governs whether a cell activates caspase-8 and dies by apoptosis.


Subject(s)
Apoptosis , Necrosis , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Animals , Caspase 8/genetics , Caspase 8/metabolism , Cell Survival , Embryo Loss , Embryonic Development , Enteritis/pathology , Fas-Associated Death Domain Protein/metabolism , Gene Knock-In Techniques , Intestine, Large/pathology , Intestine, Small/pathology , Mice , Receptor-Interacting Protein Serine-Threonine Kinases/antagonists & inhibitors , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Tumor Necrosis Factor-alpha/pharmacology
10.
Science ; 337(6101): 1541-6, 2012 Sep 21.
Article in English | MEDLINE | ID: mdl-22878500

ABSTRACT

De-ubiquitinating enzyme BAP1 is mutated in a hereditary cancer syndrome with increased risk of mesothelioma and uveal melanoma. Somatic BAP1 mutations occur in various malignancies. We show that mouse Bap1 gene deletion is lethal during embryogenesis, but systemic or hematopoietic-restricted deletion in adults recapitulates features of human myelodysplastic syndrome (MDS). Knockin mice expressing BAP1 with a 3xFlag tag revealed that BAP1 interacts with host cell factor-1 (HCF-1), O-linked N-acetylglucosamine transferase (OGT), and the polycomb group proteins ASXL1 and ASXL2 in vivo. OGT and HCF-1 levels were decreased by Bap1 deletion, indicating a critical role for BAP1 in stabilizing these epigenetic regulators. Human ASXL1 is mutated frequently in chronic myelomonocytic leukemia (CMML) so an ASXL/BAP1 complex may suppress CMML. A BAP1 catalytic mutation found in a MDS patient implies that BAP1 loss of function has similar consequences in mice and humans.


Subject(s)
Cell Transformation, Neoplastic , Genes, Tumor Suppressor , Leukemia, Myelomonocytic, Chronic/genetics , Myelodysplastic Syndromes/genetics , Tumor Suppressor Proteins/metabolism , Ubiquitin Thiolesterase/genetics , Animals , Bone Marrow Transplantation , Chromatin Immunoprecipitation , Embryonic Development , Gene Deletion , Gene Expression Regulation , Gene Knock-In Techniques , Hematopoiesis , Host Cell Factor C1/metabolism , Humans , Leukemia, Myelomonocytic, Chronic/metabolism , Leukemia, Myelomonocytic, Chronic/pathology , Mice , Mice, Knockout , Myelodysplastic Syndromes/metabolism , Myelodysplastic Syndromes/pathology , Myeloid Cells/cytology , Myeloid Cells/physiology , Myeloid Progenitor Cells/cytology , Myeloid Progenitor Cells/physiology , N-Acetylglucosaminyltransferases/metabolism , Promoter Regions, Genetic , Repressor Proteins/metabolism , Tumor Suppressor Proteins/chemistry , Tumor Suppressor Proteins/genetics , Ubiquitin Thiolesterase/chemistry , Ubiquitin Thiolesterase/metabolism
11.
PLoS One ; 7(5): e36713, 2012.
Article in English | MEDLINE | ID: mdl-22615798

ABSTRACT

The fibroblast growth factor (FGF)-FGF receptor (FGFR) signaling system plays critical roles in a variety of normal developmental and physiological processes. It is also well documented that dysregulation of FGF-FGFR signaling may have important roles in tumor development and progression. The FGFR4-FGF19 signaling axis has been implicated in the development of hepatocellular carcinomas (HCCs) in mice, and potentially in humans. In this study, we demonstrate that FGFR4 is required for hepatocarcinogenesis; the progeny of FGF19 transgenic mice, which have previously been shown to develop HCCs, bred with FGFR4 knockout mice fail to develop liver tumors. To further test the importance of FGFR4 in HCC, we developed a blocking anti-FGFR4 monoclonal antibody (LD1). LD1 inhibited: 1) FGF1 and FGF19 binding to FGFR4, 2) FGFR4-mediated signaling, colony formation, and proliferation in vitro, and 3) tumor growth in a preclinical model of liver cancer in vivo. Finally, we show that FGFR4 expression is elevated in several types of cancer, including liver cancer, as compared to normal tissues. These findings suggest a modulatory role for FGFR4 in the development and progression of hepatocellular carcinoma and that FGFR4 may be an important and novel therapeutic target in treating this disease.


Subject(s)
Carcinoma, Hepatocellular/prevention & control , Disease Models, Animal , Liver Neoplasms/prevention & control , Receptor, Fibroblast Growth Factor, Type 4/genetics , Animals , Antibodies, Neutralizing/immunology , Carcinoma, Hepatocellular/pathology , Cell Division , Liver Neoplasms/pathology , Mice , Mice, Transgenic , Receptor, Fibroblast Growth Factor, Type 4/immunology
12.
Cell ; 146(6): 918-30, 2011 Sep 16.
Article in English | MEDLINE | ID: mdl-21925315

ABSTRACT

Inhibitors of DNA binding (IDs) antagonize basic-helix-loop-helix (bHLH) transcription factors to inhibit differentiation and maintain stem cell fate. ID ubiquitination and proteasomal degradation occur in differentiated tissues, but IDs in many neoplasms appear to escape degradation. We show that the deubiquitinating enzyme USP1 promotes ID protein stability and stem cell-like characteristics in osteosarcoma. USP1 bound, deubiquitinated, and thereby stabilized ID1, ID2, and ID3. A subset of primary human osteosarcomas coordinately overexpressed USP1 and ID proteins. USP1 knockdown in osteosarcoma cells precipitated ID protein destabilization, cell-cycle arrest, and osteogenic differentiation. Conversely, ectopic USP1 expression in mesenchymal stem cells stabilized ID proteins, inhibited osteoblastic differentiation, and enhanced proliferation. Consistent with USP1 functioning in normal mesenchymal stem cells, USP1-deficient mice were osteopenic. Our observations implicate USP1 in preservation of the stem cell state that characterizes osteosarcoma and identify USP1 as a target for differentiation therapy.


Subject(s)
Endopeptidases/metabolism , Inhibitor of Differentiation Proteins/metabolism , Mesenchymal Stem Cells/cytology , Neoplastic Stem Cells/cytology , Osteosarcoma/pathology , Animals , Arabidopsis Proteins , Basic Helix-Loop-Helix Transcription Factors/metabolism , Gene Knockdown Techniques , Humans , Mice , Ubiquitin-Specific Proteases , Ubiquitination
13.
Nature ; 474(7351): 403-6, 2011 May 15.
Article in English | MEDLINE | ID: mdl-21572435

ABSTRACT

The proto-oncogenes ETV1, ETV4 and ETV5 encode transcription factors in the E26 transformation-specific (ETS) family, which includes the most frequently rearranged and overexpressed genes in prostate cancer. Despite being critical regulators of development, little is known about their post-translational regulation. Here we identify the ubiquitin ligase COP1 (also known as RFWD2) as a tumour suppressor that negatively regulates ETV1, ETV4 and ETV5. ETV1, which is mutated in prostate cancer more often, was degraded after being ubiquitinated by COP1. Truncated ETV1 encoded by prostate cancer translocation TMPRSS2:ETV1 lacks the critical COP1 binding motifs and was 50-fold more stable than wild-type ETV1. Almost all patient translocations render ETV1 insensitive to COP1, implying that this confers a selective advantage to prostate epithelial cells. Indeed, COP1 deficiency in mouse prostate elevated ETV1 and produced increased cell proliferation, hyperplasia, and early prostate intraepithelial neoplasia. Combined loss of COP1 and PTEN enhanced the invasiveness of mouse prostate adenocarcinomas. Finally, rare human prostate cancer samples showed hemizygous loss of the COP1 gene, loss of COP1 protein, and elevated ETV1 protein while lacking a translocation event. These findings identify COP1 as a tumour suppressor whose downregulation promotes prostatic epithelial cell proliferation and tumorigenesis.


Subject(s)
Nuclear Proteins/metabolism , Proto-Oncogene Proteins c-ets/metabolism , Tumor Suppressor Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , Amino Acid Motifs , Animals , Carrier Proteins/metabolism , Cell Line , Cell Line, Tumor , Cell Proliferation , Cell Transformation, Neoplastic , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Humans , Male , Mice , Nuclear Proteins/deficiency , PTEN Phosphohydrolase/deficiency , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Protein Binding , Transcription Factors/genetics , Transcription Factors/metabolism , Ubiquitin-Protein Ligases/deficiency , Ubiquitin-Protein Ligases/genetics , Ubiquitination
14.
Cancer Cell ; 19(3): 347-58, 2011 Mar 08.
Article in English | MEDLINE | ID: mdl-21397858

ABSTRACT

We screened 124 genes that are amplified in human hepatocellular carcinoma (HCC) using a mouse hepatoblast model and identified 18 tumor-promoting genes, including CCND1 and its neighbor on 11q13.3, FGF19. Although it is widely assumed that CCND1 is the main driving oncogene of this common amplicon (15% frequency in HCC), both forward-transformation assays and RNAi-mediated inhibition in human HCC cells established that FGF19 is an equally important driver gene in HCC. Furthermore, clonal growth and tumorigenicity of HCC cells harboring the 11q13.3 amplicon were selectively inhibited by RNAi-mediated knockdown of CCND1 or FGF19, as well as by an anti-FGF19 antibody. These results show that 11q13.3 amplification could be an effective biomarker for patients most likely to respond to anti-FGF19 therapy.


Subject(s)
Carcinoma, Hepatocellular/genetics , Fibroblast Growth Factors/genetics , Liver Neoplasms/genetics , Oncogene Proteins/genetics , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacology , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Chromosomes, Human, Pair 11/genetics , Cyclin D1/genetics , Cyclin D1/metabolism , Female , Fibroblast Growth Factors/immunology , Fibroblast Growth Factors/metabolism , Gene Amplification , Gene Expression Regulation, Neoplastic , Genetic Predisposition to Disease , Genomics/methods , Humans , Immunoblotting , Liver Neoplasms/drug therapy , Liver Neoplasms/pathology , Mice , Mice, Nude , Oncogene Proteins/metabolism , RNA Interference , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
15.
PLoS One ; 6(3): e17868, 2011 Mar 18.
Article in English | MEDLINE | ID: mdl-21437243

ABSTRACT

Fibroblast growth factor 19 (FGF19) is a hormone-like protein that regulates carbohydrate, lipid and bile acid metabolism. At supra-physiological doses, FGF19 also increases hepatocyte proliferation and induces hepatocellular carcinogenesis in mice. Much of FGF19 activity is attributed to the activation of the liver enriched FGF Receptor 4 (FGFR4), although FGF19 can activate other FGFRs in vitro in the presence of the coreceptor ßKlotho (KLB). In this report, we investigate the role of FGFR4 in mediating FGF19 activity by using Fgfr4 deficient mice as well as a variant of FGF19 protein (FGF19v) which is specifically impaired in activating FGFR4. Our results demonstrate that FGFR4 activation mediates the induction of hepatocyte proliferation and the suppression of bile acid biosynthesis by FGF19, but is not essential for FGF19 to improve glucose and lipid metabolism in high fat diet fed mice as well as in leptin-deficient ob/ob mice. Thus, FGF19 acts through multiple receptor pathways to elicit pleiotropic effects in regulating nutrient metabolism and cell proliferation.


Subject(s)
Bile Acids and Salts/metabolism , Fibroblast Growth Factors/pharmacology , Glucose/metabolism , Receptor, Fibroblast Growth Factor, Type 4/metabolism , Signal Transduction/drug effects , Amino Acid Sequence , Animals , Bile Acids and Salts/blood , CHO Cells , Cell Proliferation/drug effects , Cricetinae , Cricetulus , Fibroblast Growth Factors/chemistry , Glucose Tolerance Test , Hepatocytes/drug effects , Hepatocytes/metabolism , Hepatocytes/pathology , Humans , Hyperglycemia/pathology , Hyperglycemia/prevention & control , Mice , Mice, Obese , Models, Biological , Molecular Sequence Data , Recombinant Proteins/pharmacology
16.
Nat Cell Biol ; 12(4): 400-6, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20228808

ABSTRACT

The dual specificity (Tyr/Thr) phosphatase Cdc25A activates cyclin-dependent kinases (Cdks) to promote cell-cycle progression and has significant oncogenic potential. Cdc25A protein levels are regulated tightly in normal tissues, but many human cancers overexpress Cdc25A. The underlying mechanism for overexpression has been enigmatic. Here we show that Cdc25A is stabilized by the ubiquitin hydrolase Dub3. Upon binding Cdc25A, Dub3 removes the polyubiquitin modifications that mark Cdc25A for proteasomal degradation. Dub3 knockdown in cells increased Cdc25A ubiquitylation and degradation, resulting in reduced Cdk/Cyclin activity and arrest at G1/S and G2/M phases of the cell cycle. In contrast, acute Dub3 overexpression produced a signature response to oncogene induction: cells accumulated in S and G2 because of replication stress, and activated a DNA damage response. Dub3 also transformed NIH-3T3 cells and cooperated with activated H-Ras to promote growth in soft agar. Importantly, we show that Dub3 overexpression is responsible for an abnormally high level of Cdc25A in a subset of human breast cancers. Moreover, Dub3 knockdown significantly retarded the growth of breast tumour xenografts in nude mice. As a major regulator of Cdc25A, Dub3 is an example of a transforming ubiquitin hydrolase that subverts a key component of the cell cycle machinery.


Subject(s)
Breast Neoplasms/metabolism , Cell Transformation, Neoplastic/metabolism , Endopeptidases/metabolism , Oncogenes , Protein Processing, Post-Translational , cdc25 Phosphatases/metabolism , Animals , Apoptosis , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Cycle , Cell Line, Tumor , Cell Proliferation , Cell Transformation, Neoplastic/genetics , DNA Damage , Endopeptidases/genetics , Enzyme Stability , Female , Gene Expression Regulation, Neoplastic , Genes, myc , Genes, ras , Humans , Mice , Mice, Nude , NIH 3T3 Cells , Proteasome Endopeptidase Complex/metabolism , RNA Interference , Time Factors , Transfection , Transplantation, Heterologous , Tumor Burden , Ubiquitination , cdc25 Phosphatases/genetics
17.
Nature ; 463(7277): 103-7, 2010 Jan 07.
Article in English | MEDLINE | ID: mdl-20023629

ABSTRACT

MCL1 is essential for the survival of stem and progenitor cells of multiple lineages, and is unique among pro-survival BCL2 family members in that it is rapidly turned over through the action of ubiquitin ligases. B- and mantle-cell lymphomas, chronic myeloid leukaemia, and multiple myeloma, however, express abnormally high levels of MCL1, contributing to chemoresistance and disease relapse. The mechanism of MCL1 overexpression in cancer is not well understood. Here we show that the deubiquitinase USP9X stabilizes MCL1 and thereby promotes cell survival. USP9X binds MCL1 and removes the Lys 48-linked polyubiquitin chains that normally mark MCL1 for proteasomal degradation. Increased USP9X expression correlates with increased MCL1 protein in human follicular lymphomas and diffuse large B-cell lymphomas. Moreover, patients with multiple myeloma overexpressing USP9X have a poor prognosis. Knockdown of USP9X increases MCL1 polyubiquitination, which enhances MCL1 turnover and cell killing by the BH3 mimetic ABT-737. These results identify USP9X as a prognostic and therapeutic target, and they show that deubiquitinases may stabilize labile oncoproteins in human malignancies.


Subject(s)
Neoplasms/metabolism , Neoplasms/pathology , Polyubiquitin/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Ubiquitin Thiolesterase/metabolism , Animals , Apoptosis/drug effects , Biphenyl Compounds/pharmacology , Cell Line , Cell Line, Tumor , Cell Survival , DNA Damage , Docetaxel , Etoposide/pharmacology , Female , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Half-Life , Humans , Lysine/metabolism , Mice , Mice, SCID , Myeloid Cell Leukemia Sequence 1 Protein , Neoplasms/diagnosis , Nitrophenols/pharmacology , Phosphorylation/radiation effects , Piperazines/pharmacology , Prognosis , Protein Binding/radiation effects , Protein Stability , Proto-Oncogene Proteins c-bcl-2/genetics , RNA Interference , Sulfonamides/pharmacology , Taxoids/pharmacology , Ubiquitin Thiolesterase/deficiency , Ubiquitin Thiolesterase/genetics , Ubiquitination , Ultraviolet Rays , Xenograft Model Antitumor Assays
18.
Cell ; 139(2): 299-311, 2009 Oct 16.
Article in English | MEDLINE | ID: mdl-19837033

ABSTRACT

Mutations in the genes encoding the Wnt receptor Frizzled-4 (FZD4), coreceptor LRP5, or the ligand Norrin disrupt retinal vascular development and cause ophthalmic diseases. Although Norrin is structurally unrelated to Wnts, it binds FZD4 and activates the canonical Wnt pathway. Here we show that the tetraspanin Tspan12 is expressed in the retinal vasculature, and loss of Tspan12 phenocopies defects seen in Fzd4, Lrp5, and Norrin mutant mice. In addition, Tspan12 genetically interacts with Norrin or Lrp5. Overexpressed TSPAN12 associates with the Norrin-receptor complex and significantly increases Norrin/beta-catenin but not Wnt/beta-catenin signaling, whereas Tspan12 siRNA abolishes transcriptional responses to Norrin but not Wnt3A in retinal endothelial cells. Signaling defects caused by Norrin or FZD4 mutations that are predicted to impair receptor multimerization are rescued by overexpression of TSPAN12. Our data indicate that Norrin multimers and TSPAN12 cooperatively promote multimerization of FZD4 and its associated proteins to elicit physiological levels of signaling.


Subject(s)
Frizzled Receptors/metabolism , Membrane Proteins/metabolism , Receptors, G-Protein-Coupled/metabolism , Retina/embryology , Signal Transduction , beta Catenin/metabolism , Animals , Diterpenes , Endothelial Cells/metabolism , Frizzled Receptors/genetics , Humans , Mice , Receptors, G-Protein-Coupled/genetics , Tetraspanins , beta Catenin/genetics
19.
Magn Reson Med ; 62(6): 1423-30, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19859948

ABSTRACT

Whole-body MRI combined with a semiautomated hierarchical multispectral image analysis technique was evaluated as a method for detecting viable tumor tissue in a murine model of metastatic breast cancer (4T1 cell line). Whole-body apparent diffusion coefficient, T(2), and proton density maps were acquired in this study. The viable tumor tissue segmentation included three-stage k-means clustering of the parametric maps, morphologic operations, application of a size threshold, and reader discrimination of the segmented objects. The segmentation results were validated by histologic evaluation, and the detection accuracy of the technique was evaluated at three size thresholds (15, 100, and 500 voxels). The accuracy was 88.9% for a 500-voxel size threshold, and the area under receiver operating characteristic curve was 0.84. The regions of segmented viable tumor tissue within the primary tumors were found mostly on the periphery of the tumors in agreement with the histologic findings. The presented technique was found capable of detecting metastases and segmenting the viable tumor from necrotic regions within tumors found in this model. It offers a noninvasive, whole-body, viable tumor tissue detection method for preclinical and potentially clinical applications such as tumor screening and evaluating therapeutic efficacy.


Subject(s)
Algorithms , Artificial Intelligence , Breast Neoplasms/diagnosis , Image Interpretation, Computer-Assisted/methods , Magnetic Resonance Imaging/methods , Pattern Recognition, Automated/methods , Whole Body Imaging/methods , Animals , Breast Neoplasms/secondary , Cell Line, Tumor , Image Enhancement/methods , Mice , Mice, Inbred BALB C , Mice, Inbred C3H , Reproducibility of Results , Sensitivity and Specificity
20.
Cell ; 134(4): 668-78, 2008 Aug 22.
Article in English | MEDLINE | ID: mdl-18724939

ABSTRACT

Posttranslational modification of proteins with polyubiquitin occurs in diverse signaling pathways and is tightly regulated to ensure cellular homeostasis. Studies employing ubiquitin mutants suggest that the fate of polyubiquitinated proteins is determined by which lysine within ubiquitin is linked to the C terminus of an adjacent ubiquitin. We have developed linkage-specific antibodies that recognize polyubiquitin chains joined through lysine 63 (K63) or 48 (K48). A cocrystal structure of an anti-K63 linkage Fab bound to K63-linked diubiquitin provides insight into the molecular basis for specificity. We use these antibodies to demonstrate that RIP1, which is essential for tumor necrosis factor-induced NF-kappaB activation, and IRAK1, which participates in signaling by interleukin-1beta and Toll-like receptors, both undergo polyubiquitin editing in stimulated cells. Both kinase adaptors initially acquire K63-linked polyubiquitin, while at later times K48-linked polyubiquitin targets them for proteasomal degradation. Polyubiquitin editing may therefore be a general mechanism for attenuating innate immune signaling.


Subject(s)
Antibodies/metabolism , Nuclear Pore Complex Proteins/metabolism , RNA-Binding Proteins/metabolism , Ubiquitin/metabolism , Animals , Cell Line , Humans , Interleukin-1 Receptor-Associated Kinases/metabolism , Mass Spectrometry , Mice , Mice, Inbred C57BL , Nuclear Pore Complex Proteins/chemistry , Peptide Library , RNA-Binding Proteins/chemistry , Saccharomyces cerevisiae , Schizosaccharomyces , Ubiquitin/chemistry , Ubiquitination
SELECTION OF CITATIONS
SEARCH DETAIL
...